Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Type of study
Publication year range
1.
Dis Model Mech ; 17(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38616715

ABSTRACT

Preclinical and clinical studies on the administration of bone marrow-derived cells to restore perfusion show conflicting results. We conducted a systematic review and meta-analysis on preclinical studies to assess the efficacy of bone marrow-derived cells in the hind limb ischemia model and identify possible determinants of therapeutic efficacy. In vivo animal studies were identified using a systematic search in PubMed and EMBASE on 10 January 2022. 85 studies were included for systematic review and meta-analysis. Study characteristics and outcome data on relative perfusion were extracted. The pooled mean difference was estimated using a random effects model. Risk of bias was assessed for all included studies. We found a significant increase in perfusion in the affected limb after administration of bone marrow-derived cells compared to that in the control groups. However, there was a high heterogeneity between studies, which could not be explained. There was a high degree of incomplete reporting across studies. We therefore conclude that the current quality of preclinical research is insufficient (low certainty level as per GRADE assessment) to identify specific factors that might improve human clinical trials.


Subject(s)
Bone Marrow Cells , Hindlimb , Ischemia , Animals , Hindlimb/blood supply , Ischemia/therapy , Ischemia/pathology , Bone Marrow Cells/cytology , Perfusion , Bone Marrow Transplantation , Humans , Publication Bias , Cell- and Tissue-Based Therapy/methods
2.
Toxicon ; 240: 107637, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38331109

ABSTRACT

Here we describe the acute myocardial effects of an elapid (red spitting cobra, Naja pallida) and a viper (western diamondback rattlesnake, Crotalus atrox) venom using an ex vivo heart model. Our results reveal two different pathophysiological trajectories that influence heart function and morphology. While cobra venom causes a drop in contractile force, rattlesnake venom causes enhanced contractility and frequency that coincides with differences in myocellular morphology. This highlights the medical complexity of snake venom-induced cardiotoxicity.


Subject(s)
Crotalid Venoms , Naja , Venomous Snakes , Animals , Crotalus , Cardiotoxicity , Elapid Venoms/toxicity , Elapidae , Crotalid Venoms/toxicity
3.
Stem Cell Reports ; 18(11): 2222-2239, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37863044

ABSTRACT

Mechanisms that underlie homeostatic plasticity have been extensively investigated at single-cell levels in animal models, but are less well understood at the network level. Here, we used microelectrode arrays to characterize neuronal networks following induction of homeostatic plasticity in human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons co-cultured with rat astrocytes. Chronic suppression of neuronal activity through tetrodotoxin (TTX) elicited a time-dependent network re-arrangement. Increased expression of AMPA receptors and the elongation of axon initial segments were associated with increased network excitability following TTX treatment. Transcriptomic profiling of TTX-treated neurons revealed up-regulated genes related to extracellular matrix organization, while down-regulated genes related to cell communication; also astrocytic gene expression was found altered. Overall, our study shows that hiPSC-derived neuronal networks provide a reliable in vitro platform to measure and characterize homeostatic plasticity at network and single-cell levels; this platform can be extended to investigate altered homeostatic plasticity in brain disorders.


Subject(s)
Induced Pluripotent Stem Cells , Neuronal Plasticity , Humans , Rats , Animals , Cells, Cultured , Neuronal Plasticity/physiology , Neurons/metabolism , Coculture Techniques , Tetrodotoxin/pharmacology
4.
Mol Psychiatry ; 27(1): 1-18, 2022 01.
Article in English | MEDLINE | ID: mdl-33972691

ABSTRACT

Activity in the healthy brain relies on a concerted interplay of excitation (E) and inhibition (I) via balanced synaptic communication between glutamatergic and GABAergic neurons. A growing number of studies imply that disruption of this E/I balance is a commonality in many brain disorders; however, obtaining mechanistic insight into these disruptions, with translational value for the patient, has typically been hampered by methodological limitations. Cadherin-13 (CDH13) has been associated with autism and attention-deficit/hyperactivity disorder. CDH13 localizes at inhibitory presynapses, specifically of parvalbumin (PV) and somatostatin (SST) expressing GABAergic neurons. However, the mechanism by which CDH13 regulates the function of inhibitory synapses in human neurons remains unknown. Starting from human-induced pluripotent stem cells, we established a robust method to generate a homogenous population of SST and MEF2C (PV-precursor marker protein) expressing GABAergic neurons (iGABA) in vitro, and co-cultured these with glutamatergic neurons at defined E/I ratios on micro-electrode arrays. We identified functional network parameters that are most reliably affected by GABAergic modulation as such, and through alterations of E/I balance by reduced expression of CDH13 in iGABAs. We found that CDH13 deficiency in iGABAs decreased E/I balance by means of increased inhibition. Moreover, CDH13 interacts with Integrin-ß1 and Integrin-ß3, which play opposite roles in the regulation of inhibitory synaptic strength via this interaction. Taken together, this model allows for standardized investigation of the E/I balance in a human neuronal background and can be deployed to dissect the cell-type-specific contribution of disease genes to the E/I balance.


Subject(s)
Cadherins , GABAergic Neurons , Parvalbumins , Cadherins/metabolism , GABAergic Neurons/metabolism , Humans , Induced Pluripotent Stem Cells , Integrins/metabolism , Parvalbumins/metabolism , Synapses/metabolism
5.
Neurobiol Dis ; 163: 105587, 2022 02.
Article in English | MEDLINE | ID: mdl-34923109

ABSTRACT

Monoamine neurotransmitter abundance affects motor control, emotion, and cognitive function and is regulated by monoamine oxidases. Among these, Monoamine oxidase A (MAOA) catalyzes the degradation of dopamine, norepinephrine, and serotonin into their inactive metabolites. Loss-of-function mutations in the X-linked MAOA gene have been associated with Brunner syndrome, which is characterized by various forms of impulsivity, maladaptive externalizing behavior, and mild intellectual disability. Impaired MAOA activity in individuals with Brunner syndrome results in bioamine aberration, but it is currently unknown how this affects neuronal function, specifically in dopaminergic (DA) neurons. Here we generated human induced pluripotent stem cell (hiPSC)-derived DA neurons from three individuals with Brunner syndrome carrying different mutations and characterized neuronal properties at the single cell and neuronal network level in vitro. DA neurons of Brunner syndrome patients showed reduced synaptic density but exhibited hyperactive network activity. Intrinsic functional properties and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated synaptic transmission were not affected in DA neurons of individuals with Brunner syndrome. Instead, we show that the neuronal network hyperactivity is mediated by upregulation of the GRIN2A and GRIN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), resulting in increased NMDAR-mediated currents. By correcting a MAOA missense mutation with CRISPR/Cas9 genome editing we normalized GRIN2A and GRIN2B expression, NMDAR function and neuronal population activity to control levels. Our data suggest that MAOA mutations in Brunner syndrome increase the activity of dopaminergic neurons through upregulation of NMDAR function, which may contribute to the etiology of Brunner syndrome associated phenotypes.


Subject(s)
Disruptive, Impulse Control, and Conduct Disorders/genetics , Dopaminergic Neurons/metabolism , Genetic Diseases, X-Linked/genetics , Intellectual Disability/genetics , Monoamine Oxidase/deficiency , Monoamine Oxidase/genetics , Mutation , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/metabolism , Aggression , Disruptive, Impulse Control, and Conduct Disorders/metabolism , Disruptive, Impulse Control, and Conduct Disorders/physiopathology , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/physiopathology , Humans , Induced Pluripotent Stem Cells , Intellectual Disability/metabolism , Intellectual Disability/physiopathology , Male , Monoamine Oxidase/metabolism , Nerve Net/metabolism , Nerve Net/physiopathology , Synapses/metabolism , Synaptic Transmission/genetics
6.
Front Mol Neurosci ; 14: 772000, 2021.
Article in English | MEDLINE | ID: mdl-34803610

ABSTRACT

Posttranslational modification of histones and related gene regulation are shown to be affected in an increasing number of neurological disorders. SETD1A is a chromatin remodeler that influences gene expression through the modulation of mono- di- and trimethylation marks on Histone-H3-Lysine-4 (H3K4me1/2/3). H3K4 methylation is predominantly described to result in transcriptional activation, with its mono- di- and trimethylated forms differentially enriched at promoters or enhancers. Recently, dominant mostly de novo variants in SETD1A have clinically been linked to developmental delay, intellectual disability (DD/ID), and schizophrenia (SCZ). Affected individuals often display both developmental and neuropsychiatric abnormalities. The primary diagnoses are mainly dependent on the age at which the individual is assessed. Investigations in mouse models of SETD1A dysfunction have been able to recapitulate key behavioral features associated with ID and SCZ. Furthermore, functional investigations suggest disrupted synaptic and neuronal network function in these mouse models. In this review, we provide an overview of pre-clinical studies on the role of SETD1A in neuronal development. A better understanding of the pathobiology underlying these disorders may provide novel opportunities for therapeutic intervention. As such, we will discuss possible strategies to move forward in elucidating the genotype-phenotype correlation in SETD1A associated disorders.

7.
Nat Commun ; 10(1): 4928, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666522

ABSTRACT

Kleefstra syndrome (KS) is a neurodevelopmental disorder caused by mutations in the histone methyltransferase EHMT1. To study the impact of decreased EHMT1 function in human cells, we generated excitatory cortical neurons from induced pluripotent stem (iPS) cells derived from KS patients. Neuronal networks of patient-derived cells exhibit network bursting with a reduced rate, longer duration, and increased temporal irregularity compared to control networks. We show that these changes are mediated by upregulation of NMDA receptor (NMDAR) subunit 1 correlating with reduced deposition of the repressive H3K9me2 mark, the catalytic product of EHMT1, at the GRIN1 promoter. In mice EHMT1 deficiency leads to similar neuronal network impairments with increased NMDAR function. Finally, we rescue the KS patient-derived neuronal network phenotypes by pharmacological inhibition of NMDARs. Summarized, we demonstrate a direct link between EHMT1 deficiency and NMDAR hyperfunction in human neurons, providing a potential basis for more targeted therapeutic approaches for KS.


Subject(s)
Craniofacial Abnormalities/genetics , Heart Defects, Congenital/genetics , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cerebral Cortex/cytology , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Craniofacial Abnormalities/metabolism , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Heart Defects, Congenital/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Induced Pluripotent Stem Cells , Intellectual Disability/metabolism , Loss of Function Mutation , Male , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Primary Cell Culture , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Up-Regulation
8.
Brain Struct Funct ; 223(9): 4211-4226, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30187194

ABSTRACT

Heterozygous mutations of the Forkhead-box protein 2 (FOXP2) gene in humans cause childhood apraxia of speech. Loss of Foxp2 in mice is known to affect striatal development and impair motor skills. However, it is unknown if striatal excitatory/inhibitory balance is affected during development and if the imbalance persists into adulthood. We investigated the effect of reduced Foxp2 expression, via a loss-of-function mutation, on striatal medium spiny neurons (MSNs). Our data show that heterozygous loss of Foxp2 decreases excitatory (AMPA receptor-mediated) and increases inhibitory (GABA receptor-mediated) currents in D1 dopamine receptor positive MSNs of juvenile and adult mice. Furthermore, reduced Foxp2 expression increases GAD67 expression, leading to both increased presynaptic content and release of GABA. Finally, pharmacological blockade of inhibitory activity in vivo partially rescues motor skill learning deficits in heterozygous Foxp2 mice. Our results suggest a novel role for Foxp2 in the regulation of striatal direct pathway activity through managing inhibitory drive.


Subject(s)
Corpus Striatum/physiology , Excitatory Postsynaptic Potentials , Forkhead Transcription Factors/physiology , Inhibitory Postsynaptic Potentials , Neurons/physiology , Repressor Proteins/physiology , gamma-Aminobutyric Acid/physiology , Animals , Forkhead Transcription Factors/genetics , Glutamate Decarboxylase/metabolism , Learning/physiology , Mice, Inbred C57BL , Mice, Transgenic , Motor Skills , Receptors, Dopamine D1/physiology , Repressor Proteins/genetics , Synapses/physiology
9.
J Vis Exp ; (119)2017 01 08.
Article in English | MEDLINE | ID: mdl-28117798

ABSTRACT

Neurons derived from human induced Pluripotent Stem Cells (hiPSCs) provide a promising new tool for studying neurological disorders. In the past decade, many protocols for differentiating hiPSCs into neurons have been developed. However, these protocols are often slow with high variability, low reproducibility, and low efficiency. In addition, the neurons obtained with these protocols are often immature and lack adequate functional activity both at the single-cell and network levels unless the neurons are cultured for several months. Partially due to these limitations, the functional properties of hiPSC-derived neuronal networks are still not well characterized. Here, we adapt a recently published protocol that describes production of human neurons from hiPSCs by forced expression of the transcription factor neurogenin-212. This protocol is rapid (yielding mature neurons within 3 weeks) and efficient, with nearly 100% conversion efficiency of transduced cells (>95% of DAPI-positive cells are MAP2 positive). Furthermore, the protocol yields a homogeneous population of excitatory neurons that would allow the investigation of cell-type specific contributions to neurological disorders. We modified the original protocol by generating stably transduced hiPSC cells, giving us explicit control over the total number of neurons. These cells are then used to generate hiPSC-derived neuronal networks on micro-electrode arrays. In this way, the spontaneous electrophysiological activity of hiPSC-derived neuronal networks can be measured and characterized, while retaining interexperimental consistency in terms of cell density. The presented protocol is broadly applicable, especially for mechanistic and pharmacological studies on human neuronal networks.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/metabolism , Microarray Analysis , Neurons/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cellular Reprogramming , Fibroblasts/cytology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Lentivirus/genetics , Microelectrodes , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Front Psychol ; 6: 1816, 2015.
Article in English | MEDLINE | ID: mdl-26635706

ABSTRACT

Speech requires precise motor control and rapid sequencing of highly complex vocal musculature. Despite its complexity, most people produce spoken language effortlessly. This is due to activity in distributed neuronal circuitry including cortico-striato-thalamic loops that control speech-motor output. Understanding the neuro-genetic mechanisms involved in the correct development and function of these pathways will shed light on how humans can effortlessly and innately use spoken language and help to elucidate what goes wrong in speech-language disorders. FOXP2 was the first single gene identified to cause speech and language disorder. Individuals with FOXP2 mutations display a severe speech deficit that includes receptive and expressive language impairments. The neuro-molecular mechanisms controlled by FOXP2 will give insight into our capacity for speech-motor control, but are only beginning to be unraveled. Recently FOXP2 was found to regulate genes involved in retinoic acid (RA) signaling and to modify the cellular response to RA, a key regulator of brain development. Here we explore evidence that FOXP2 and RA function in overlapping pathways. We summate evidence at molecular, cellular, and behavioral levels that suggest an interplay between FOXP2 and RA that may be important for fine motor control and speech-motor output. We propose RA signaling is an exciting new angle from which to investigate how neuro-genetic mechanisms can contribute to the (spoken) language ready brain.

SELECTION OF CITATIONS
SEARCH DETAIL
...